Download as pdf or txt
Download as pdf or txt
You are on page 1of 18

RESEARCH ARTICLES

Cite as: T. Nakamura et al., Science


10.1126/science.aaw0843 (2020).

Noncanonical transnitrosylation network contributes to


synapse loss in Alzheimer’s disease
Tomohiro Nakamura1,2*†, Chang-ki Oh1,2†, Lujian Liao1‡, Xu Zhang1,2, Kevin M. Lopez2, Daniel Gibbs3, Amanda
K. Deal1, Henry R. Scott1, Brian Spencer3, Eliezer Masliah3§, Robert A. Rissman3,4, John R. Yates III1, Stuart A.
Lipton1,2,3*
1Departments of Molecular Medicine and Neuroscience, and Neuroscience Translational Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
2Neurodegenerative Disease Center, Scintillon Institute, San Diego, CA 92121, USA. 3Department of Neurosciences, University of California, San Diego, School of Medicine,
La Jolla, CA 92093, USA. 4VA San Diego Healthcare System, San Diego, CA, USA.
*Corresponding author. Email: tnakamura@scripps.edu (T.N.); slipton@scripps.edu (S.A.L.)
†These authors contributed equally to this work. ‡Present address: Shanghai Key Laboratory of Regulatory Biology and Shanghai Key Laboratory of Brain Functional

Downloaded from http://science.sciencemag.org/ on December 4, 2020


Genomics, School of Life Sciences, East China Normal University, Shanghai 200241, China. §Present address: National Institute on Aging/NIH, Bethesda, MD, USA.

We describe mechanistically-distinct enzymes, i.e., a kinase, a guanosine triphosphatase and a ubiquitin


protein hydrolase, which function in disparate biochemical pathways, that can also act in concert to
mediate a series of redox reactions. Each enzyme manifests a second, noncanonical function –
transnitrosylation – triggering a pathological biochemical cascade in Alzheimer’s disease (AD). The
resulting series of transnitrosylation reactions contributes to synapse loss, the major pathological
correlate to cognitive decline in AD. We conclude that enzymes with distinct primary reaction mechanisms
can form a completely separate network for aberrant transnitrosylation. This network operates in the post-
reproductive period, so natural selection against such abnormal activity may be decreased.

Excessive nitric oxide (NO) can engender nitrosative stress in biochemical pathways can form a transnitrosylation cascade
the nervous system, contributing to neurodegenerative dam- that contributes to synapse loss in AD. We demonstrate that
age (1, 2). In Alzheimer’s disease (AD), oligomerized amyloid- the deubiquitinylating enzyme, Uch-L1 (Ubiquitin carboxyl-
β peptide (Aβ), neuronal hyperexcitability, and neuroinflam- terminal hydrolase isozyme-L1), contributes to a transnitro-
mation associated with aging can each trigger NO generation sylation cascade leading to synaptic damage in AD that in-
and resultant S-nitrosylation (donation of an NO group most volves transfer of a NO group from Uch-L1 to Cdk5 and then
likely as a nitrosonium cation intermediate, NO+) to specific to Drp1. Although other potential members of the cascade re-
cysteine thiol (or more properly thiolate anion, -S−) (3–14). main to be determined, our results show pathways mediating
One such cysteine is located on dynamin-related protein 1 a series of aberrant transnitrosylation reactions may be im-
(Drp1), a guanosine triphosphatase (GTPase) that mediates portant in the etiology of neurodegenerative disorders.
mitochondrial fission (15). This posttranslational redox reac-
tion, forming SNO-Drp1, hyperactivates the enzyme, leading S-Nitrosylation of Uch-L1 in cell lines and AD
to mitochondrial fragmentation and bioenergetic compro- transgenic mice by biotin switch and mass spectrometry
mise, with consequent synapse loss, in part because of the We examined upstream events to S-nitrosylation of Cdk5
great demand for energy during neurotransmission (15, 16). (forming SNO-Cdk5), which in turn leads to SNO-Drp1
Because reduction in synapse number is closely correlated formation and consequent Aβ-related synaptic spine loss in
with cognitive decline in AD (17, 18), this nitrosylation reac- AD (15, 20). The ubiquitin hydrolase Uch-L1 can potentiate
tion may contribute to disease pathogenesis (4). S-Nitrosyl- cyclin-dependent kinase activity by an unknown reaction
ated cyclin-dependent kinase 5 (Cdk5) (19) acts to pass NO to mechanism not involving its ubiquitin hydrolase activity (21),
Drp1 to form SNO-Drp1 via a reaction termed transnitrosyla- and we found that this mechanism involves transnitrosyla-
tion (20). However, additional members of this pathological tion to Cdk5.
cascade are yet to be determined. Characterizing this aber- Aberrant S-nitrosylation of a number of proteins that
rant series of reactions may further reveal its contribution to affect their canonical enzymatic activity has been demon-
the pathogenesis of AD and possible therapeutic implications strated in AD and other neurologic diseases (4), including
of the pathway. We used both in vitro and in vivo models of members of the ubiquitin pathway such as the E3 ligase
AD as well as human AD postmortem brains to show that en- Parkin and the ubiquitin hydrolase Uch-L1 (22–24). However,
zymes with disparate catalytic activities from different we discovered that aberrant and seemingly unrelated

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 1
S-nitrosylated enzymes are linked by multiple transnitrosyla- (to reductively remove NO from SNO-protein) resulted in an
tion steps between them that result in a wholly disparate cas- increase in the active form of Uch-L1, consistent with increased
cade of biochemical pathological activity. We exposed human ubiquitinylation activity due to denitrosylation of Uch-L1.
neural SH-SY5Y cells to a physiological NO donor, S-nitro- Decreased Uch-L1 activity is observed in human brains
socysteine (SNOC). We then made cell lysates and analyzed with AD as well as in AD mouse models (28, 29). Hence,
them for S-nitrosylated Uch-L1. Using the biotin-switch SNO-Uch-L1 formation in AD brain, with consequent inhibi-
method (25), we found evidence for the formation of SNO- tion of Uch-L1 activity, might contribute to this effect. Over-
Uch-L1 under these conditions (Fig. 1A). Additionally, endog- expression of Uch-L1 decreases Aβ production, increases
enous NO, generated by activation of stably-expressed neu- activity of protein kinase A (PKA) and cAMP response
ronal NO synthase (nNOS) in human embryonic kidney element-binding protein (CREB), and improves synaptic
(HEK) cells (designated HEK-nNOS cells), also resulted in function and memory performance in AD transgenic mice
formation of SNO-Uch-L1 (Fig. 1B); nNOS activation by the (29–31). Therefore, Uch-L1 activity appears to be important
calcium ionophore A23187 led to a ~2-fold increase in the rel- in the pathogenesis of the disease.
ative abundance of SNO-Uch-L1 in replicate experiments. The

Downloaded from http://science.sciencemag.org/ on December 4, 2020


absence of ascorbate in the reaction mixture abrogated detec- Oligomeric Aβ triggers SNO-Uch-L1 formation and
tion of the increase in SNO-Uch-L1 via the biotin-switch assay synapse loss in AD models in vitro and in vivo
because, under these conditions, NO was not removed and We reasoned that if the therapeutic action of Uch-L1 overex-
replaced by biotin (Fig. 1, A and B). Of potential pathological pression was mediated by decreasing endogenous Aβ produc-
relevance, we found increased SNO-Uch-L1 compared to wild- tion, then the pathological effects on synapses of adding
type (WT) littermates in cerebrocortices of AD transgenic exogenous Aβ oligomers might not be greatly affected by
mouse models overexpressing human amyloid precursor pro- Uch-L1 overexpression. On the other hand, if the beneficial
tein (hAPP-J20 and Tg2576), which produce Aβ oligomers effect of Uch-L1 occurred, at least in part, through another
(Fig. 1C and fig. S1). The very high abundance of Uch-L1 in function of the enzyme, we should observe this therapeutic
the brain might favor its S-nitrosylation, not necessarily re- action in the presence of exogenous Aβ oligomers. Indeed,
quiring a catalyst for the reaction. S-nitrosylation of Uch-L1 occurred in a neuronal cell-based
We used site-directed mutagenesis in HEK-293 cells to in- model upon exposure to Aβ oligomers–incubation of primary
vestigate which cysteine residue was S-nitrosylated in Uch- rat cerebrocortical cultures in 500 nM Aβ1-42 oligomers
L1. Mutation of Cys152 decreased formation of SNO-Uch-L1, resulted in formation of SNO-Uch-L1 as determined by the
indicating that it is a major site of S-nitrosylation after expo- biotin-switch assay (Fig. 3A and fig. S3A).
sure to exogenous SNOC (Fig. 1D) or endogenous NO in HEK- Additionally, after exposure of rat cerebrocortical neurons
nNOS cells (Fig. 1E). Additionally, the NOS inhibitor NG-nitro- to 500 nM Aβ1-42 oligomers, approximately half of the
l-arginine (NNA) prevented S-nitrosylation of WT Uch-L1, dendritic spines were lost (Fig. 3, B and C) (15, 20, 32–34);
confirming that NO was indeed the reactant (Fig. 1E). We loss of synaptic spines was dependent on NO, as preincuba-
used mass spectrometry-based S-nitrosoproteomics analysis tion in NNA abrogated the effect (15, 20). Transfection with
to confirm the predominant site of S-nitrosylation as Cys152 non-nitrosylatable mutant Uch-L1, but not with WT Uch-L1,
(Fig. 2, A and B, and table S1). Moreover, we identified an decreased the Aβ-induced loss of dendritic spines in a statis-
acidic (Glu)-basic (Arg) region as a candidate motif for S-ni- tically significant manner, although there was also a trend
trosylation surrounding the critical Cys residue in the atomic toward improvement with WT Uch-L1 (Fig. 3C and fig. S3B).
resolution model of Uch-L1 (Fig. 2C) (12, 26, 27). This trend with WT Uch-L1 may reflect overexpression of WT
protein, which can act as a sink for NO (allowing substantial
Effect of S-nitrosylation on Uch-L1 deubiquitinylation WT protein to remain un-nitrosylated) and therefore poten-
activity in cell-based models and AD transgenic mice tially offer some benefit (15). However, non-nitrosylatable
To determine the effect of formation of SNO-Uch-L1, we mutant protein is more effective and showed statistically sig-
monitored Uch-L1 binding to ubiquitin and Uch-L1 deubiqui- nificant synaptic protection.
tinylation activity. Exposure to SNOC decreased Uch-L1 asso- To determine if S-nitrosylated Uch-L1 also contributes in
ciated with ubiquitin and decreased deubiquitinylation vivo to synapse loss, we used the hAPP-J20 transgenic mouse
activity in both recombinant and cell-based assays (fig. S2, A model of AD, which overexpresses oligomeric Aβ in the brain
to C). As a control, non-nitrosylatable mutant Uch-L1(C152S) (35). We generated lentiviral vectors expressing tdTomato
was not affected by SNOC. Additionally, in the hAPP-J20 fluorescent protein and Uch-L1 (WT or C152S), and injected
transgenic mouse model of AD, we observed a decrease in the them into the dentate gyrus of hAPP-J20 AD mice (fig. S4, A
active form of Uch-L1, i.e., Uch-L1 bound to ubiquitin on im- and B). Two months later, we evaluated the histological ef-
munoblots (fig. S2D). Conversely, treatment with ascorbate fects on presynaptic integrity. Quantitative unbiased confocal

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 2
immunohistochemistry of hippocampal sections with an an- occurred, but SNOC also S-nitrosylated Cdk5 and Drp1. The
tibody to the presynaptic marker synaptophysin (SY38) (36, fact that knockdown of Uch-L1 decreased to a statistically sig-
37) revealed that expression of Uch-L1(C152S), but not that of nificant degree the formation of SNO-Cdk5 (at 10 min) and
WT Uch-L1, significantly increased SY38 signal compared to SNO-Drp1 (at 10 min and even more so at 25 min) under these
that obtained from vehicle-treated hAPP-J20 AD mice (Fig. 3, conditions argues that SNO-Uch-L1 triggered downstream
D and E). Although overexpression of WT protein afforded a transnitrosylation reactions (20, 39).
trend toward presynaptic protection, probably because it By graphing the difference of relative SNO-Cdk5 and SNO-
acted as a sink for NO (15), only non-nitrosylatable mutant Drp1 concentrations, as calculated from control (siCTL) val-
protein resulted in statistically significant protection of the ues compared to those in cells lacking Uch-L1 (siUch-L1), we
presynaptic marker. These findings are consistent with the can subtract the effect of S-nitrosylation due to SNOC and
notion that SNO-Uch-L1 contributes to synapse loss in AD quantify the effect of transnitrosylation between the enzymes
models. Because synapse loss is a close neuropathological (Fig. 4, C and D). The temporal delay in appearance of SNO-
correlate to cognitive decline in human AD, this finding sup- Drp1 relative to SNO-Cdk5 is consistent with the notion that
ports biological relevance to the disease state. SNO-Cdk5, in turn, may transnitrosylate Drp1.

Downloaded from http://science.sciencemag.org/ on December 4, 2020


To characterize this putative transnitrosylation pathway
Transnitrosylation from Uch-L1 to Cdk5 to Drp1 from SNO-Cdk5 to Drp1, we used in vitro nitrosylation assays.
Aβ oligomer-induced S-nitrosylation of Drp1 results in We transfected SH-SY5Y neural cells with WT Uch-L1 or mu-
mitochondrial fragmentation, bioenergetic compromise, and tant Uch-L1(C152S), each tagged with V5 (derived from a
consequent synapse loss (15, 16, 38). Studies with purified small epitope found in paramyxovirus proteins), prepared
recombinant proteins support direct transfer of an NO group cell lysates, and performed immunoprecipitation with anti-
(representing transnitrosylation) to Drp1 from SNO-Cdk5 body to V5. Immunoprecipitated Uch-L1 protein (WT or
(20), although the upstream effector linking Aβ-induced NO C152S mutant) was then exposed to SNOC and incubated for
generation to S-nitrosylation of Cdk5 was unknown. Given a sufficient period of time to ensure near total dissipation of
our new evidence for S-nitrosylation of Uch-L1 in AD trans- NO from SNOC (20, 39). The WT or C152S mutant Uch-L1 im-
genic mouse brain and the fact that Uch-L1 potentiates munoprecipitates were then added to cell lysates containing
cyclin-dependent kinase activity by a reaction mechanism hemagglutinin (HA)-tagged Cdk5 (Fig. 5, A to E). We moni-
not involving its ubiquitin hydrolase activity (21), we tested tored possible transnitrosylation reactions by probing for for-
whether Uch-L1 might contribute to an NO-mediated path- mation of SNO-V5-Uch-L1, SNO-HA-Cdk5, and endogenous
way to synapse loss, leading to a series of transnitrosylation SNO-Drp1 by biotin-switch assay. SNO-Cdk5 and SNO-Drp1
steps among these enzymes from diverse biochemical path- formed in the WT Uch-L1-immunoprecipitated lysates after
ways (NO group transfer from Uch-L1 to Cdk5 to Drp1). SNOC exposure but significantly less was found in immuno-
We performed kinetic, cell-based S-nitrosylation assays to precipitates from mutant Uch-L1(C152S) (Fig. 5, A to E). Alt-
monitor possible transfer of an NO group from SNO-Uch-L1 hough immunoprecipitate experiments do not rule out the
to Cdk5 and then to Drp1. Using the biotin-switch assay, we possibility that additional members of the cascade participate
found evidence for formation of both SNO-Uch-L1 and SNO- in these transnitrosylation reactions, they support participa-
Cdk5 in HEK-293 cells within 10 min of exposure to SNOC; tion of Uch-L1 and Cdk5 in the reaction sequence leading to
with increasing formation of SNO-Drp1 by 25 min, concur- the generation of SNO-Drp1.
rently with a relative decrease in the amount of SNO-Cdk5 The relative degree of transnitrosylation of Drp1 resulting
(Fig. 4, A to D). The fact that RNAi depletion of Uch-L1, com- from S-nitrosylated Uch-L1 was dependent on the amount
pared to control siRNA (siCTL), partially prevented for- of endogenous Cdk5. When Cdk5 was depleted by immunopre-
mation of SNO-Cdk5 under these conditions at 10 min is cipitation, the amount of transnitrosylated SNO-Drp1 de-
consistent with the notion that transnitrosylation occurs creased (Fig. 5, F to I). The degree of immunodepletion of
from upstream SNO-Uch-L1 to Cdk5 (Fig. 4, A to C). Similarly, endogenous Cdk5 corresponded with the decrement in SNO-
RNAi depletion of Uch-L1 also partially prevented the subse- Drp1 formation in the presence of WT Uch-L1 but not in the
quent formation of SNO-Drp1 (Fig. 4D). Although RNAi presence of C152S mutant Uch-L1 (Fig. 5G). This finding
depletion of Uch-L1 decreased SNO-Cdk5 and SNO-Drp1 gen- confirms that SNO-Cdk5, rather than SNO-Uch-L1, predomi-
eration, it did not eliminate it, presumably because SNOC is nantly mediates transnitrosylation to Drp1 under these condi-
a non-enzymatic donor of NO, producing super-physiological tions. This type of experiment is also consistent with the notion
amounts of protein S-nitrosylation that would be expected to that these enzymes transnitrosylate with a preference of one
continue undeterred when competing with transnitrosyla- substrate over another. Mechanistically, the particular envi-
tion among proteins. To clarify further, this experiment indi- ronment of the thiolate anion can kinetically favor one NO
cates that donation of NO from SNOC to SNO-Uch-L1 donor over another.

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 3
In cell lysates expressing either HA-Cdk5 or V5-Uch-L1, of their respective Nernst equations:
we found transnitrosylation from SNO-Uch-L1 to Cdk5, and
0′ 0′ RT  [ UchL1SNO ][ Cdk5red ] 
EUchL1 − ECdk5 =
− ⋅ ln  74.02 mV
=
then to Drp1, but lack of NO group transfer from SNO-Cdk5
zF  [ UchL1 ][ Cdk5 ] 
to Uch-L1 (Fig. 6A). These experiments, although lacking  red SNO 

formal derivation of rate constants, turnover number (kcat) (1)


or substrate affinity (inversely related to Km) to support an  [ UchL1red ][ Cdk5SNO ] 
∆G 0′ =
− RT ⋅ ln  −18.35 kJ/mol
=
enzymatic process (40), do provide additional evidence for  [ UchL1 ][ Cdk5 ] 
specific transnitrosylation reactions because of relative  SNO red 

substrate preference. Thus, the entire pathway of NO transfer (2)


from SNO-Uch-L1 to SNO-Cdk5 to SNO-Drp1 had to be where “SNO” represents the S-nitrosylated (or oxidized) pro-
present in order to observe significant SNO-Drp1 formation tein and “red” represents the chemically reduced protein, as
under these conditions. Nonetheless, other proteins present obtained from densitometric analysis of redox immunoblots
in the immunoprecipitates may participate in this series of (Fig. 6B). Because the Nernst equation uses log values, the
transnitrosylation events. To show more definitively that difference in Nernst equations represents the natural log of
the ratio of effective concentrations of the reduced and oxi-

Downloaded from http://science.sciencemag.org/ on December 4, 2020


SNO-Uch-L1 directly transfers NO to Cdk5, we performed
an in vitro assay with purified recombinant proteins. dized forms of the protein. Thus, the absolute concentrations
SNO-Uch-L1 could directly transfer an NO group to Cdk5 of the reduced protein and oxidized (in this case, S-nitrosyl-
(fig. S5). ated) protein are not needed for the evaluation of the blots or
Because of differences in methods between standard im- for the resulting calculation; only the relative ratio is re-
munoblotting and the biotin-switch method, one cannot quired. Hence, we can treat the reactions quantitatively with-
compare the density of the bands representing the total input out actually knowing the exact protein concentrations.
protein and the S-nitrosylated protein for evaluation of the Using this method, we found that transnitrosylation from
absolute concentration of the SNO-protein. However, we can SNO-Uch-L1 to Cdk5 is predicted to be energetically very fa-
compare the ratio of SNO-protein to total (input) protein to vorable under steady-state conditions in intact cells. Evidence
obtain the relative amount of S-nitrosylated protein, and we for this was obtained by obtaining the relative redox potential
can then compare this ratio across various preparations (20, for the reaction (Eq. 1, ΔE0′ = 74.02 ± 4.60 mV [n = 4]), which
39, 41). Therefore, more critical to the overall hypothesis of a in turn allowed us to calculate the associated change in Gibbs
series of transnitrosylation events than the actual magnitude free energy (Eq. 2, ΔG0′ = −18.35 ± 1.14 kJ/mol [n = 4]), pre-
of an individual SNO-protein band generated is the fact that dicting the reaction will proceed spontaneously. Our previous
we could detect significant differences in their relative abun- results indicated that the subsequent transnitrosylation from
dance. This ratio concept is also amenable to precise quanti- SNO-Cdk5 to Drp1 was also energetically favorable (20). Col-
tative analysis of electron transfer and associated energetics lectively, these findings lead us to propose that an NO group
of transnitrosylation reactions under steady-state conditions, is transferred from SNO-Uch-L1 to Cdk5, which leads to sub-
as described below. sequent transfer from SNO-Cdk5 to Drp1 in an energetically-
favorable manner.
Nernst equations to predict transnitrosylation
pathways and Gibbs free energy at steady state S-Nitrosylation of Uch-L1 in human AD brains
In general, biological redox reactions often do not go to com- To assess the potential pathophysiological relevance of our
pletion and depend on the relative redox potential of the pair findings in human AD, we obtained human postmortem
of proteins that interact. Thus, SNO-protein bands observed brain tissue (table S2). Abundance of SNO-UchL1 was signif-
in biotin-switch assays are not expected to be completely icantly increased in human brains with advanced stages of
abated when individual upstream or downstream compo- AD compared to that in control brains (Fig. 7A and fig. S6).
nents are disrupted. Accordingly, we devised a quantitative Amounts of SNO-Uch-L1 were extremely low by these meth-
method based on the Nernst equation to characterize these ods in control human brains, consistent with the notion that
transnitrosylation reactions thermodynamically (20, 39). For S-nitrosylation of Uch-L1 represents an aberrant nitrosyla-
this analysis, we make the assumption that these reactions go tion event occurring in the diseased state only. To determine
to steady state, as might be reasonably expected to occur in a whether accumulation of SNO-Uch-L1 in human AD brain
chronic disease. The Nernst equation quantifies the electro- might be of pathophysiological significance, we calculated
motive force for net electron movement between a redox pair. the ratio of SNO-Uch-L1 (by biotin-switch assay) to total Uch-
To calculate the difference in the electromotive force (ΔE) be- L1 (as quantified from immunoblots), using methods previ-
tween two redox pairs, e.g., for transnitrosylation between ously described (20, 39, 41). In human AD brain this ratio was
Uch-L1 and Cdk5, we calculated the difference in the values comparable to or greater than that encountered in our

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 4
neuronal cell-based models exhibiting Aβ/SNO-Uch-L1— In conclusion, we have discovered sequential NO transfer
induced dendritic spine loss (Figs. 3A and 7B) or in the brains among unrelated enzymes that is not accounted for by the
of hAPP-J20 AD mice (Figs. 1C and 7B), which exhibit synapse canonical catalytic activity originally discovered for the en-
loss (42). Taken together, these findings indicated that zymes. This concept of “noncanonical transnitrosylation net-
the aberrant transnitrosylation cascade from SNO-Uch-L1 works” may represent a set of biochemical reactions not
to Cdk5 to Drp1 may contribute to the synaptic pathology currently enveloped in standard bioinformatics packages
observed in AD (Fig. 7C). such as gene ontology (GO) enrichment or KEGG (Kyoto En-
cyclopedia of Genes and Genomes) pathway analysis, and
Discussion thus this finding could change our concept of pathway anal-
The abundance of Uch-L1 protein is decreased in human ysis and selection pressure on such cascades. Although it is
brains with sporadic AD (28). On the other hand, overexpres- unusual for a deleterious biochemical reaction network to de-
sion of Uch-L1 delays AD progression in vivo in animal mod- velop because of natural selection, the fact that these aber-
els (29). This is thought to occur, at least in part, because of rant reactions occur in the setting of neurodegenerative
decreased Aβ production (30, 31). In the continued presence diseases in aged individuals, far beyond their reproductive

Downloaded from http://science.sciencemag.org/ on December 4, 2020


of exogenous oligomeric Aβ, however, Uch-L1 still modulates years, may explain why they are subject to less evolutionary
synaptic damage in AD models, implicating a second mecha- selection pressure (47).
nism of Uch-L1 action. Along these lines, in in vivo experi- Our mass spectrometry screening method uncovered a
ments, expression of mutant Uch-L1(C152S) in mice is large number of potentially S-nitrosylated or transnitrosyl-
neuroprotective (43), implicating the possible importance of ated proteins in the brain (table S1). In the future, as mass
redox reactions at the critical Cys residue 152 of Uch-L1. spectrometry methods continue to advance, additional mem-
We found that Cys152 of Uch-L1 is S-nitrosylated in human bers of this noncanonical transnitrosylation network and ad-
AD brain to a similar degree as in our cell-based and animal ditional transnitrosylation pathways in various disease states
models of AD. This reaction forms SNO-Uch-L1, which in may be discovered. In summary, we find that enzymes previ-
turn, transnitrosylates Cdk5, an enzyme that functions ously thought to be in distinct biochemical cascades are ac-
in neuronal maturation and migration, among other pro- tually linked by pathological crosstalk via transnitrosylation
cesses. Next, SNO-Cdk5 transnitrosylates Drp1, a GTPase that under disease conditions that may contribute in a causal
mediates mitochondrial fission. Not only do these transnitro- manner to the disease process. The transnitrosylation activity
sylation reactions occur in cell-based immunoprecipitates, of these enzymes may represent a therapeutic target for AD.
but also in in vitro transnitrosylation assays using puri-
fied/recombinant enzymes, showing that the donation of Materials and methods
the NO group proceeds directly from one protein to another. Cell lines
Hence, the data show that there is overall donation of an NO SH-SY5Y cells (ATCC; CRL-2266) and HEK cells stably ex-
group via a series transnitrosylation reactions from Uch-L1 pressing nNOS (HEK-nNOS cells, gift from Drs. David Bredt
to Cdk5 to Drp1 (Fig. 7C). The S-nitrosylation of Drp1 triggers and Solomon Snyder) were maintained in DMEM (Thermo
excessive mitochondrial fission/fragmentation with conse- Fisher Scientific) supplemented with 10% fetal bovine serum,
quent bioenergetic loss and synaptic damage (15, 20). This 2 mM l-glutamine (Thermo Fisher Scientific), 50 IU/ml peni-
finding may influence progression of AD, as synapse loss is cillin (Omega Scientific), and 50 μg/ml streptomycin (Omega
a major neuropathological correlate of cognitive decline in Scientific). For HEK-nNOS cell culture, 100 μg/ml geneticin
AD (17, 18). (Thermo Fisher Scientific) was also included in the culture
The proteins that participate in this transnitrosylation medium to maintain the expression of nNOS, and nNOS ac-
cascade (Uch-L1/Cdk5/Drp1) also function in totally dispar- tivation was achieved by the incubation with the calcium ion-
ate pathways with different mechanisms of enzymatic ophore, A23187 (10 μM, EMD Millipore). Primary rat cortical
action (ubiquitin hydrolase, kinase, and GTPase, respec- neurons were isolated and cultured as described previously
tively). One reason that this second noncanonical function of (15, 39, 41, 48).
transnitrosylation may occur is that the catalytic sites of
many types of enzymes contain cysteine residues, and their E. coli strain for purification of recombinant Uch-L1
thiol groups (in fact, thiolates) are nucleophiles that can BL21 (DE3) E. coli (Agilent Technology) were transformed
perform a reversible nucleophilic attack on the nitroso nitro- with a pET28a vector containing His-tagged Uch-L1 (WT or
gen via an S-nitrosylation reaction scheme (44–46). This C152S mutant) and grown in 2XYT medium until the optical
reaction is particularly favored under conditions of high density measured at 600 nm (OD 600) reached 0.5. Expres-
NO-induced nitrosative stress, as found in neurodegenerative sion of Uch-L1 was induced by 0.5 mM IPTG for 16 hours, and
diseases associated with aging. E. coli were collected by centrifugation. The bacterial pellets

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 5
were re-suspended in PBS, frozen down at −80°C for 20 min, vector (Thermo Fisher Scientific). For knockdown experi-
and then thawed at room temperature for cell disruption. ments, HEK-293 cells were transfected with siUch-L1 (Am-
Cells were further disrupted by sonication on ice, followed by bion, #4390824, ID: s14616) or Silencer Select Negative
incubation in 1% Triton X-100 at 4°C for 30 min. The samples Control No. 1 siRNA (Ambion, #4390843) using Lipofec-
were centrifuged for 15 min at 10,000 × g, collected as super- tamine RNAiMAX Transfection Reagent (Thermo Fisher Sci-
natants, and then centrifuged again to completely remove entific) according to the manufacturer’s protocol and
bacterial debris. Recombinant Uch-L1 protein was purified incubated for 72 hours to achieve Uch-L1 knockdown. For
using Ni-NTA agarose (Qiagen) or TALON metal affinity resin bacterial expression, the Uch-L1 sequence was inserted into
(Takara) at room temperature for 1 hour. Imidazole (50 mM) pET28a, adding a His tag to its N terminus. C47S, C90S,
was used for protein elution, and excess imidazole removed C132S, C152S, C201S, C220S Uch-L1 mutants were generated
by PD-10 columns (GE Healthcare Life Sciences). using the QuikChange Lightning Multi Site-Directed Muta-
genesis Kit (Agilent Technologies) according to the manufac-
Animals and patient brain samples turer’s protocol.
We used two mouse models of AD (3-6 months old): 1)

Downloaded from http://science.sciencemag.org/ on December 4, 2020


Tg2576, overexpressing a mutant form of human (h)APP with Biotin-switch assays and immunoblots
the Swedish mutation (KM670/671NL) (Taconic Biosciences, For analysis of S-nitrosylated proteins, we performed the bi-
Model 1349) (49), and 2) hAPP-J20, expressing hAPP with two otin-switch assay as previously described (15, 39, 41). In brief,
familial AD-linked mutations (the Swedish and Indiana cell or tissue homogenates were incubated with the MMTS
[V717F] mutations) (The Jackson Laboratory, 34836-JAX) (S-methyl methanethiosulfonate, Sigma-Aldrich) blocking
(42). Age and sex matched samples were randomly chosen buffer (10 mM MMTS, 2.5% SDS in HEN [100 mM Hepes, pH
and assigned to different experimental groups. All experi- 7.4, 1 mM EDTA, 0.1 mM neocuproine] buffer) to block free
ments were performed in accordance with the guidelines out- thiol groups. After specific reduction of S-nitrosothiols by 20
lined by the Institutional Animal Care and Use Committees mM ascorbate, the newly generated free thiol groups were bi-
at The Sanford Burnham Prebys Medical Discovery Institute, otinylated with 1 mM biotin-HPDP (Thermo Fisher Scientific
the Scintillon Institute, or The Scripps Research Institute. or Soltec Ventures). For control groups, ascorbate was omit-
Postmortem brain tissues from AD or age-matched con- ted from the procedure to ensure the specificity of biotinyla-
trol patients (age <95) were collected at the University of Cal- tion. The biotinylated proteins were pulled down using High
ifornia, San Diego, School of Medicine from subjects whose Capacity NeutrAvidin agarose (Thermo Fisher Scientific),
age, postmortem interval, and gender are described in table and the purified proteins were subjected to immunoblot anal-
S2. Human brain samples were analyzed with institutional ysis. For the loading control, 5% input was saved before con-
permission under the state of California and NIH guidelines. ducting a NeutrAvidin pull-down.
Informed consent was obtained according to procedures ap- Protein samples were resolved by Bolt Bis-Tris Plus gels
proved by the Institutional Review Board at the University of (Thermo Fisher Scientific) and transferred to Immobilon-FL
California, San Diego, School of Medicine. PVDF membranes (EMD Millipore). After blocking with Od-
yssey TBS blocking buffer (Li-Cor), the membranes were in-
NO donors, Aβ oligomer preparation, cell cultures, cubated with primary antibodies against Uch-L1 (1:1000,
plasmids, and transfections Sigma-Aldrich, U5258), myc (1:1000, Cell Signaling Technol-
For induction of S-nitrosothiol formation using exogenous ogy, 2276), V5 (1:5000, Thermo Fisher Scientific, R960-25),
NO donors, the rapidly-decaying NO donor, SNOC was syn- HA (1:1000, Santa Cruz Biotechnology, sc-805), Cdk5 (1:1000,
thesized as described previously (50) and employed for short- Santa Cruz Biotechnology, sc-173 or Cell Signaling Technol-
term additions of NO. Oligomerized Aβ1-42 was prepared as we ogy, 12134), or Drp1 (1:1000, BD Biosciences, 611113). IR-dye
described previously (36). HEK-nNOS cells and rat primary 680LT-conjugated goat anti-mouse IgG (1:15000, Li-Cor, 926-
neurons were transfected with Lipofectamine 2000 (Thermo 68020) and IR-dye 800CW-conjugated goat anti-rabbit IgG
Fisher Scientific) as per the manufacturer’s instruction. (1:15000, Li-Cor, 926-32211) were used as secondary antibod-
Lipofectamine LTX with PLUS reagent (Thermo Fisher Sci- ies. For visualization of target proteins, the membranes were
entific) was used for transfection of SH-SY5Y cells. Measure- scanned with an infrared-based Odyssey imaging system (Li-
ments of dendritic spine density in rat primary cortical Cor), and quantification of each band was performed with
neuron cultures were performed in a masked fashion, as we Image Studio software (Li-Cor).
previously described (15, 36, 48).
For exogenous expression of Uch-L1 in mammalian cells, Resin-assisted capture of S-nitrosylated proteins
the human cDNA sequence for Uch-L1 containing N-terminal (SNO-RAC) for unbiased mass spectrometry analysis
V5 tag or C-terminal myc tag was subcloned into pcDNA3.1 To purify S-nitrosylated proteins, including SNO-Uch-L1,

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 6
SNO-RAC was performed as previously described (51) with Ubiquitin pull-down assays
minor modifications. In brief, mouse brain cerebrocortical ly- To prepare resin-bound SNO-Uch-L1 in vitro, recombinant
sates were prepared in cold HENTS (1% Triton X-100, 0.1% Uch-L1 bound to Ni-NTA agarose was mixed with SNOC (100
SDS in HEN [100 mM Hepes, pH 7.4, 1 mM EDTA, 0.1 mM μM) or “old” SNOC (from which NO had been dissipated), and
neocuproine]) buffer, assayed for protein concentration us- left at room temperature for 30 min in the dark with occa-
ing the Bio-Rad Protein Assay Dye Reagent Concentrate (Bi- sional mixing. The samples were washed once with PBS, and
oRad), and diluted with HEN buffer to a concentration of 1 resuspended in PBS containing 0.5% Triton X-100. Recombi-
μg/μl. SNOC (final concentration 25 μM) was added to 250 μl nant Ubiquitin (Ub) (Sigma) was then mixed with resin-
of mouse brain lysate, and the mixture was left at room tem- bound SNO-Uch-L1 (or a reduced form of Uch-L1) and incu-
perature for 25 min in the dark to allow formation of S-nitro- bated at room temperature for 1 hour with rotation. After in-
sylated (SNO)-proteins. Protein thiols were blocked with cubation, the resin was washed twice with PBS containing
MMTS (50 mM) at 50°C for 30 min, and excess MMTS re- 0.5% Triton X-100. The samples were resuspended in Nu-
moved by acetone precipitation. S-Nitrosothiols were then PAGE LDS sample buffer (Thermo Fisher Scientific), boiled
converted to free thiols with ascorbate (50 mM), and then at 95°C for 3 min, and loaded onto NuPAGE Bis-Tris gel. The

Downloaded from http://science.sciencemag.org/ on December 4, 2020


captured by thiopropyl sepharose 6B resin (GE Healthcare bands were visualized by the standard Coomassie staining
Life Sciences) with rotation at room temperature for 2 hours. method.
The samples were washed twice with HENS buffer (HEN con-
taining 1% SDS), twice with HENS/NaCl (HENS containing Uch-L1 deubiquitinylation activity assays
400 mM NaCl), twice with HENS/10 (1:10 HENS), and then Deubiquitinylation activity of recombinant Uch-L1 was fluo-
once with trypsin digestion buffer (50 mM NH4HCO3, 1 mM rometrically assayed using Ub-7-amido-4-methylcoumarin
EDTA). Trypsin digestion was conducted on resin at 37°C for (Ub-AMC, Boston Biochem) as described (29) with minor
12 hours, and the eluate was subjected to analysis by mass modification. In brief, 0.25 μM Ub-AMC and 0.02 μM Uch-L1
spectrometry (MS). (or SNO-Uch-L1) were mixed in a final volume of 100 μl in
Digested peptide mixtures were pressure-loaded onto a reaction buffer (50 mM Tris-HCl, pH 7.6, 0.5 mM EDTA), and
fused silica capillary column of 250-μm internal diameter incubated for 10 min at 25°C. The amount of AMC released
(I.D.) packed with 3 cm of 5 μm Partisphere strong cation ex- from the Ub-AMC substrate was monitored using SpectraMax
changer (SCX, Whatman) and 3 cm of 10 μm Jupiter reversed- M2 (Molecular Devices) with excitation and emission wave-
phase C12 material (Phenomenex); the SCX was end fritted lengths of 380 nm and 460 nm, respectively.
with immobilized Kasil 1624 (PQ Corp.). After desalting, a 100 Activity of Uch-L1 in cells or tissue lysates was assessed
μm I.D. capillary with a 5 μm pulled tip packed with 15 cm of with the activity-based probe, ubiquitin-vinylmethyl ester
4 μm Jupiter C12 material was attached to a zero-dead-vol- (Ub-VME), irreversibly modifying the “active” form of ubiq-
ume union, and the entire split-column was placed in line uitin C‐terminal hydrolases and thus causing a shift in elec-
with an Eksigent nano HPLC pump (Eksigent) and analyzed trophoretic mobility on immunoblots (55, 56). For the Ub-
using modified, four-step multi-dimensional protein identifi- VME assay, cell or tissue lysate was freshly prepared in PBS
cation technology (MudPIT), as described previously (52). containing 0.1% Triton X-100. The reaction was initiated by
The eluted peptides were electrosprayed directly into a hy- incubating 2 μg of lysate with 0.2 μM Ub-VME (Boston Bio-
brid LTQ-Orbitrap mass spectrometer (ThermoFinnigan) chem), incubated for 5 min at room temperature, and
with a distal spray voltage of 2.4 kV. A cycle of one full-scan stopped by adding NuPAGE LDS sample buffer (Thermo
mass spectrum (400-1400 m/z) followed by 6 data-dependent Fisher Scientific). Proteins were resolved by NuPAGE Bis-Tris
MS/MS scans were repeated continuously. Mass spectrum gel followed by Western blotting.
data were collected using Thermo XCalibur 3.1 (Thermo
Fisher Scientific). The MS/MS spectra were searched with the Lentivirus production, stereotaxic injection, and
ProLucid (a SEQUEST-based software) algorithm (53) against quantitative immunohistochemistry
the EBI mouse IPI database concatenated to a decoy database Uch-L1 (WT or C152S mutant) was subcloned into lentiviral
in which the sequence for each entry in the original database pCDH-EF1-dTomato-T2A-Kpn21 (System Biosciences) and
was reversed. No modification on cysteine was allowed to re- used for preparation of active lentiviral particles for animal
flect the reduced SNO thiols. The search results were assem- injections (57). hAPP-J20 AD transgenic mice or non-trans-
bled and filtered using the DTASelect program (54) with a genic littermate control mice at the age of 3-4 months re-
peptide false-positive rate of 1%. All peptides identified were ceived bilateral, stereotactic injections of lentiviral vectors
at least half tryptic, and precursor ion mass tolerance was set into the dentate gyrus at a rate of 1 μl per 2 min for a total of
to 20 parts per million (ppm). 2 μl of lentivirus infused into each hemisphere. Coordinates
used for viral injection were as follows (35): AP-2.1, ML±1.8,

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 7
and DV-2.0. Eight weeks after lentiviral injection, mice were “old” SNOC and then mixed with SH-SY5Y cell lysates immu-
anesthetized, perfused transcardially with saline (0.9% NaCl) nodepleted of Cdk5. Transnitrosylation was assessed by bio-
containing 20 mU/ml Heparin (Sigma), followed by 4% PFA tin-switch assay. To calculate the redox potential and change
perfusion. Brains were then removed and fixed in 4% PFA for in Gibbs free energy for the transnitrosylation reaction, Uch-
48 hours at 4°C and kept in 1% PFA until analyzed. Quantita- L1-V5 and CDK5-HA co-transfected SH-SY5Y cells were ex-
tive immunohistochemistry was performed as described (36, posed to 50 μM SNOC; after 30 min, cell lysates were sub-
37). Immunolabeling was performed using mouse monoclo- jected to biotin-switch assay in the absence or presence of
nal antibody against Synaptophysin (Clone SY38, Millipore). MMTS free-thiol blocking agent, allowing us to observe total
Analysis of co-localization was performed on the whole den- protein (both the reduced and S-nitrosylated forms) and oxi-
tate gyrus image using 3 separate images per condition by Fiji dized protein (the S-nitrosylated forms), respectively. Subse-
(ImageJ) software using the Squassh plugin as previously de- quently, immunoblotted bands were quantified by
scribed (58, 59). All procedures described here were approved densitometry, and the values used in a ratio of Nernst equa-
by the Institutional Animal Care and Use Committees at the tions, as described in the manuscript text.
Scintillon Institute and The Scripps Research Institute, For in vitro transnitrosylation assays with recombinant

Downloaded from http://science.sciencemag.org/ on December 4, 2020


where the work was performed. proteins to detect direct transfer of the NO group from one
protein to another, we first exposed purified Uch-L1 (10 μM)
Transnitrosylation assays to 200 μM SNOC and then incubated for 30 min to generate
SH-SY5Y cells were transfected with Uch-L1-V5 or Cdk5-HA. SNO-Uch-L1 while allowing NO to dissipate from SNOC. Re-
After 1 day, Uch-L1-V5 expressing cells were lysed in RIPA sidual SNOC or cysteine remaining after NO dissipation was
buffer (50 mM Tris–HCl, pH 8.0, 150 mM NaCl, 1% Nonidet removed with a Zeba Spin Desalting Column (Thermo Fisher
P-40, 0.5% deoxycholate, 0.1% sodium dodecyl sulfate) and Scientific), and the resulting purified SNO-Uch-L1 was incu-
immunoprecipitated overnight at 4°C with anti-V5 antibody bated with recombinant Cdk5/p25 (2 μM, Abcam, ab60761)
conjugated to Dynabeads Protein G (Thermo Fisher Scien- for an additional 45 min to facilitate transnitrosylation from
tific), and then washed 3 times with high-salt RIPA buffer (50 Uch-L1 to Cdk5. Note that p25 was included for stabilization
mM Tris–HCl, pH 8.0, 500 mM NaCl, 1% Nonidet P-40, 0.5% of Cdk5, as we and others had previously shown that Cdk5
deoxycholate, 0.1% sodium dodecyl sulfate). Immunoprecipi- was the S-nitrosylated species (19). The biotin-switch assay
tated Uch-L1-V5 was exposed to decayed or freshly prepared was then performed as described above.
100 μM SNOC in lysis buffer (PBS containing 0.1% Triton X-
100) for 30 min at room temperature in the dark with gentle Quantification and statistical analysis
shaking. Concurrently, cell lysates were prepared in lysis buffer Fiji (ImageJ) and Image Studio were used for quantification
(PBS containing 1% Triton X-100) from SH-SY5Y cells express- of immunohistochemistry and immunoblot data, respec-
ing Cdk5-HA. Immunoprecipitated Uch-L1-V5 exposed to tively. A Power Analysis of our prior data was used to deter-
SNOC (or as a control “old” SNOC, from which NO had been mine the number of replicates needed for statistical purposes.
dissipated) was then added to SH-SY5Y whole-cell lysates and The number of replicates or experiments are indicated in the
incubated for 1 hour at room temperature in the dark with gen- individual figure legends. Data are expressed as mean + SEM.
tle shaking. These reaction mixtures were subsequently sub- Differences between experimental groups were evaluated us-
jected to biotin-switch assay after addition of MMTS blocking ing an ANOVA followed by a post hoc test for multiple com-
buffer. Dynabeads used for immunoprecipitation were re- parisons (Dunnett’s or Bonferroni’s) or a Student’s t test for
moved by magnet rack prior to acetone precipitation. comparison between two groups. A P value  < 0.05 was con-
For transnitrosylation assays using Cdk5-immuno- sidered significant. Statistical analyses were performed using
depleted cell lysates, SH-SY5Y cells were transfected with WT GraphPad Prism software.
Uch-L1-V5, mutant Uch-L1(C152S)-V5, or Drp1-HA. After 1
REFERENCES AND NOTES
day, Uch-L1-V5—expressing cells were lysed in lysis buffer 1. V. L. Dawson, T. M. Dawson, E. D. London, D. S. Bredt, S. H. Snyder, Nitric oxide
(PBS containing 1% Triton X-100) and immunoprecipitated mediates glutamate neurotoxicity in primary cortical cultures. Proc. Natl. Acad.
with anti-V5 antibody. SH-SY5Y cells expressing Drp1-HA Sci. U.S.A. 88, 6368–6371 (1991). doi:10.1073/pnas.88.14.6368 Medline
were also lysed in lysis buffer and immunodepleted overnight 2. M. R. Hara, N. Agrawal, S. F. Kim, M. B. Cascio, M. Fujimuro, Y. Ozeki, M. Takahashi,
J. H. Cheah, S. K. Tankou, L. D. Hester, C. D. Ferris, S. D. Hayward, S. H. Snyder, A.
at 4°C with anti-Cdk5 antibody (C-8, Santa Cruz Biotechnol- Sawa, S-nitrosylated GAPDH initiates apoptotic cell death by nuclear
ogy, sc-173); or rabbit IgG (Santa Cruz, SC-2027) was used as translocation following Siah1 binding. Nat. Cell Biol. 7, 665–674 (2005).
a control antibody. Immunoprecipitated Uch-L1-V5 was doi:10.1038/ncb1268 Medline
washed in high-salt RIPA buffer (50 mM Tris–HCl, pH 8.0, 3. S. A. Lipton, Y.-B. Choi, Z.-H. Pan, S. Z. Lei, H.-S. V. Chen, N. J. Sucher, J. Loscalzo,
D. J. Singel, J. S. Stamler, A redox-based mechanism for the neuroprotective and
500 mM NaCl, 1% Nonidet P-40, 0.5% deoxycholate, 0.1% so- neurodestructive effects of nitric oxide and related nitroso-compounds. Nature
dium dodecyl sulfate) and exposed to freshly prepared or 364, 626–632 (1993). doi:10.1038/364626a0 Medline

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 8
4. T. Nakamura, S. Tu, M. W. Akhtar, C. R. Sunico, S. Okamoto, S. A. Lipton, Aberrant T. M. Dawson, S-nitrosylation of parkin regulates ubiquitination and compromises
protein s-nitrosylation in neurodegenerative diseases. Neuron 78, 596–614 parkin’s protective function. Science 304, 1328–1331 (2004).
(2013). doi:10.1016/j.neuron.2013.05.005 Medline doi:10.1126/science.1093891 Medline
5. D. T. Hess, A. Matsumoto, S. O. Kim, H. E. Marshall, J. S. Stamler, Protein S- 23. D. Yao, Z. Gu, T. Nakamura, Z.-Q. Shi, Y. Ma, B. Gaston, L. A. Palmer, E. M.
nitrosylation: Purview and parameters. Nat. Rev. Mol. Cell Biol. 6, 150–166 (2005). Rockenstein, Z. Zhang, E. Masliah, T. Uehara, S. A. Lipton, Nitrosative stress linked
doi:10.1038/nrm1569 Medline to sporadic Parkinson’s disease: S-nitrosylation of parkin regulates its E3
6. L. J. Ignarro, Nitric oxide. A novel signal transduction mechanism for transcellular ubiquitin ligase activity. Proc. Natl. Acad. Sci. U.S.A. 101, 10810–10814 (2004).
communication. Hypertension 16, 477–483 (1990). doi:10.1161/01.HYP.16.5.477 doi:10.1073/pnas.0404161101 Medline
Medline 24. R. Kumar, D. K. Jangir, G. Verma, S. Shekhar, P. Hanpude, S. Kumar, R. Kumari, N.
7. U. Seneviratne, A. Nott, V. B. Bhat, K. C. Ravindra, J. S. Wishnok, L.-H. Tsai, S. R. Singh, N. Sarovar Bhavesh, N. Ranjan Jana, T. Kanti Maiti, S-nitrosylation of
Tannenbaum, S-nitrosation of proteins relevant to Alzheimer’s disease during UCHL1 induces its structural instability and promotes α-synuclein aggregation.
early stages of neurodegeneration. Proc. Natl. Acad. Sci. U.S.A. 113, 4152–4157 Sci. Rep. 7, 44558 (2017). doi:10.1038/srep44558 Medline
(2016). doi:10.1073/pnas.1521318113 Medline 25. S. R. Jaffrey, H. Erdjument-Bromage, C. D. Ferris, P. Tempst, S. H. Snyder, Protein
8. P. T. Doulias, M. Tenopoulou, J. L. Greene, K. Raju, H. Ischiropoulos, Nitric oxide S-nitrosylation: A physiological signal for neuronal nitric oxide. Nat. Cell Biol. 3,
regulates mitochondrial fatty acid metabolism through reversible protein S- 193–197 (2001). doi:10.1038/35055104 Medline
nitrosylation. Sci. Signal. 6, rs1 (2013). doi:10.1126/scisignal.2003252 Medline 26. C. T. Stomberski, D. T. Hess, J. S. Stamler, Protein S-nitrosylation: Determinants
9. G. Hao, B. Derakhshan, L. Shi, F. Campagne, S. S. Gross, SNOSID, a proteomic of specificity and enzymatic regulation of S-nitrosothiol-based signaling. Antioxid.
method for identification of cysteine S-nitrosylation sites in complex protein Redox Signal. 30, 1331–1351 (2019). doi:10.1089/ars.2017.7403 Medline

Downloaded from http://science.sciencemag.org/ on December 4, 2020


mixtures. Proc. Natl. Acad. Sci. U.S.A. 103, 1012–1017 (2006). 27. J. S. Stamler, E. J. Toone, S. A. Lipton, N. J. Sucher, (S)NO signals: Translocation,
doi:10.1073/pnas.0508412103 Medline regulation, and a consensus motif. Neuron 18, 691–696 (1997).
10. T. S. Wijasa, M. Sylvester, N. Brocke-Ahmadinejad, S. Schwartz, F. Santarelli, V. doi:10.1016/S0896-6273(00)80310-4 Medline
Gieselmann, T. Klockgether, F. Brosseron, M. T. Heneka, Quantitative proteomics 28. J. Choi, A. I. Levey, S. T. Weintraub, H. D. Rees, M. Gearing, L.-S. Chin, L. Li,
of synaptosome S-nitrosylation in Alzheimer’s disease. J. Neurochem. 152, 710– Oxidative modifications and down-regulation of ubiquitin carboxyl-terminal
726 (2020). doi:10.1111/jnc.14870 Medline hydrolase L1 associated with idiopathic Parkinson’s and Alzheimer’s diseases. J.
11. Y. I. Lee, D. Giovinazzo, H. C. Kang, Y. Lee, J. S. Jeong, P.-T. Doulias, Z. Xie, J. Hu, Biol. Chem. 279, 13256–13264 (2004). doi:10.1074/jbc.M314124200 Medline
M. Ghasemi, H. Ischiropoulos, J. Qian, H. Zhu, S. Blackshaw, V. L. Dawson, T. M. 29. B. Gong, Z. Cao, P. Zheng, O. V. Vitolo, S. Liu, A. Staniszewski, D. Moolman, H.
Dawson, Protein microarray characterization of the S-nitrosoproteome. Mol. Cell. Zhang, M. Shelanski, O. Arancio, Ubiquitin hydrolase Uch-L1 rescues β-amyloid-
Proteomics 13, 63–72 (2014). doi:10.1074/mcp.M113.032235 Medline induced decreases in synaptic function and contextual memory. Cell 126, 775–
12. J. Jia, A. Arif, F. Terenzi, B. Willard, E. F. Plow, S. L. Hazen, P. L. Fox, Target-selective 788 (2006). doi:10.1016/j.cell.2006.06.046 Medline
protein S-nitrosylation by sequence motif recognition. Cell 159, 623–634 (2014). 30. M. Zhang, Y. Deng, Y. Luo, S. Zhang, H. Zou, F. Cai, K. Wada, W. Song, Control of
doi:10.1016/j.cell.2014.09.032 Medline BACE1 degradation and APP processing by ubiquitin carboxyl-terminal hydrolase
13. S. M. Marino, V. N. Gladyshev, Structural analysis of cysteine S-nitrosylation: A L1. J. Neurochem. 120, 1129–1138 (2012). doi:10.1111/j.1471-4159.2011.07644.x
modified acid-based motif and the emerging role of trans-nitrosylation. J. Mol. Medline
Biol. 395, 844–859 (2010). doi:10.1016/j.jmb.2009.10.042 Medline 31. M. Zhang, F. Cai, S. Zhang, S. Zhang, W. Song, Overexpression of ubiquitin
14. M. D. Kornberg, N. Sen, M. R. Hara, K. R. Juluri, J. V. K. Nguyen, A. M. Snowman, L. carboxyl-terminal hydrolase L1 (UCHL1) delays Alzheimer’s progression in vivo.
Law, L. D. Hester, S. H. Snyder, GAPDH mediates nitrosylation of nuclear proteins. Sci. Rep. 4, 7298 (2014). doi:10.1038/srep07298 Medline
Nat. Cell Biol. 12, 1094–1100 (2010). doi:10.1038/ncb2114 Medline 32. G. M. Shankar, B. L. Bloodgood, M. Townsend, D. M. Walsh, D. J. Selkoe, B. L.
15. D. H. Cho, T. Nakamura, J. Fang, P. Cieplak, A. Godzik, Z. Gu, S. A. Lipton, S- Sabatini, Natural oligomers of the Alzheimer amyloid-β protein induce reversible
nitrosylation of Drp1 mediates β-amyloid-related mitochondrial fission and synapse loss by modulating an NMDA-type glutamate receptor-dependent
neuronal injury. Science 324, 102–105 (2009). doi:10.1126/science.1171091 signaling pathway. J. Neurosci. 27, 2866–2875 (2007).
Medline doi:10.1523/JNEUROSCI.4970-06.2007 Medline
16. T. Nakamura, P. Cieplak, D. H. Cho, A. Godzik, S. A. Lipton, S-nitrosylation of Drp1 33. B. Calabrese, G. M. Shaked, I. V. Tabarean, J. Braga, E. H. Koo, S. Halpain, Rapid,
links excessive mitochondrial fission to neuronal injury in neurodegeneration. concurrent alterations in pre- and postsynaptic structure induced by naturally-
Mitochondrion 10, 573–578 (2010). doi:10.1016/j.mito.2010.04.007 Medline secreted amyloid-β protein. Mol. Cell. Neurosci. 35, 183–193 (2007).
17. S. T. DeKosky, S. W. Scheff, Synapse loss in frontal cortex biopsies in Alzheimer’s doi:10.1016/j.mcn.2007.02.006 Medline
disease: Correlation with cognitive severity. Ann. Neurol. 27, 457–464 (1990). 34. W. Wei, L. N. Nguyen, H. W. Kessels, H. Hagiwara, S. Sisodia, R. Malinow, Amyloid
doi:10.1002/ana.410270502 Medline β from axons and dendrites reduces local spine number and plasticity. Nat.
18. R. D. Terry, E. Masliah, D. P. Salmon, N. Butters, R. DeTeresa, R. Hill, L. A. Hansen, Neurosci. 13, 190–196 (2010). doi:10.1038/nn.2476 Medline
R. Katzman, Physical basis of cognitive alterations in Alzheimer’s disease: 35. M. Cissé, B. Halabisky, J. Harris, N. Devidze, D. B. Dubal, B. Sun, A. Orr, G. Lotz, D.
Synapse loss is the major correlate of cognitive impairment. Ann. Neurol. 30, 572– H. Kim, P. Hamto, K. Ho, G.-Q. Yu, L. Mucke, Reversing EphB2 depletion rescues
580 (1991). doi:10.1002/ana.410300410 Medline cognitive functions in Alzheimer model. Nature 469, 47–52 (2011).
19. P. Zhang, P.-C. Yu, A. H. K. Tsang, Y. Chen, A. K. Y. Fu, W.-Y. Fu, K. K. Chung, N. Y. doi:10.1038/nature09635 Medline
Ip, S-nitrosylation of cyclin-dependent kinase 5 (cdk5) regulates its kinase activity 36. M. Talantova, S. Sanz-Blasco, X. Zhang, P. Xia, M. W. Akhtar, S. Okamoto, G.
and dendrite growth during neuronal development. J. Neurosci. 30, 14366–14370 Dziewczapolski, T. Nakamura, G. Cao, A. E. Pratt, Y.-J. Kang, S. Tu, E. Molokanova,
(2010). doi:10.1523/JNEUROSCI.3899-10.2010 Medline S. R. McKercher, S. A. Hires, H. Sason, D. G. Stouffer, M. W. Buczynski, J. P.
20. J. Qu, T. Nakamura, G. Cao, E. A. Holland, S. R. McKercher, S. A. Lipton, S- Solomon, S. Michael, E. T. Powers, J. W. Kelly, A. Roberts, G. Tong, T. Fang-
Nitrosylation activates Cdk5 and contributes to synaptic spine loss induced by β- Newmeyer, J. Parker, E. A. Holland, D. Zhang, N. Nakanishi, H.-S. V. Chen, H.
amyloid peptide. Proc. Natl. Acad. Sci. U.S.A. 108, 14330–14335 (2011). Wolosker, Y. Wang, L. H. Parsons, R. Ambasudhan, E. Masliah, S. F. Heinemann, J.
doi:10.1073/pnas.1105172108 Medline C. Piña-Crespo, S. A. Lipton, Aβ induces astrocytic glutamate release,
21. T. Kabuta, T. Mitsui, M. Takahashi, Y. Fujiwara, C. Kabuta, C. Konya, Y. Tsuchiya, Y. extrasynaptic NMDA receptor activation, and synaptic loss. Proc. Natl. Acad. Sci.
Hatanaka, K. Uchida, H. Hohjoh, K. Wada, Ubiquitin C-terminal hydrolase L1 (UCH- U.S.A. 110, E2518–E2527 (2013). doi:10.1073/pnas.1306832110 Medline
L1) acts as a novel potentiator of cyclin-dependent kinases to enhance cell 37. S. A. Lipton, T. Rezaie, A. Nutter, K. M. Lopez, J. Parker, K. Kosaka, T. Satoh, S. R.
proliferation independently of its hydrolase activity. J. Biol. Chem. 288, 12615– McKercher, E. Masliah, N. Nakanishi, Therapeutic advantage of pro-electrophilic
12626 (2013). doi:10.1074/jbc.M112.435701 Medline drugs to activate the Nrf2/ARE pathway in Alzheimer’s disease models. Cell Death
22. K. K. Chung, B. Thomas, X. Li, O. Pletnikova, J. C. Troncoso, L. Marsh, V. L. Dawson, Dis. 7, e2499 (2016). doi:10.1038/cddis.2016.389 Medline

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 9
38. M. J. Barsoum, H. Yuan, A. A. Gerencser, G. Liot, Y. Kushnareva, S. Gräber, I. 55. A. Borodovsky, B. M. Kessler, R. Casagrande, H. S. Overkleeft, K. D. Wilkinson, H.
Kovacs, W. D. Lee, J. Waggoner, J. Cui, A. D. White, B. Bossy, J.-C. Martinou, R. J. L. Ploegh, A novel active site-directed probe specific for deubiquitylating enzymes
Youle, S. A. Lipton, M. H. Ellisman, G. A. Perkins, E. Bossy-Wetzel, Nitric oxide- reveals proteasome association of USP14. EMBO J. 20, 5187–5196 (2001).
induced mitochondrial fission is regulated by dynamin-related GTPases in doi:10.1093/emboj/20.18.5187 Medline
neurons. EMBO J. 25, 3900–3911 (2006). doi:10.1038/sj.emboj.7601253 Medline 56. A. E. Cartier, S. N. Djakovic, A. Salehi, S. M. Wilson, E. Masliah, G. N. Patrick,
39. T. Nakamura, L. Wang, C. C. L. Wong, F. L. Scott, B. P. Eckelman, X. Han, C. Regulation of synaptic structure by ubiquitin C-terminal hydrolase L1. J. Neurosci.
Tzitzilonis, F. Meng, Z. Gu, E. A. Holland, A. T. Clemente, S. Okamoto, G. S. 29, 7857–7868 (2009). doi:10.1523/JNEUROSCI.1817-09.2009 Medline
Salvesen, R. Riek, J. R. Yates 3rd, S. A. Lipton, Transnitrosylation of XIAP regulates 57. J. D. Reuter, X. Fang, C. S. Ly, K. K. Suter, D. Gibbs, Assessment of hazard risk
caspase-dependent neuronal cell death. Mol. Cell 39, 184–195 (2010). associated with the intravenous use of viral vectors in rodents. Comp. Med. 62,
doi:10.1016/j.molcel.2010.07.002 Medline 361–370 (2012). Medline
40. D. A. Mitchell, M. A. Marletta, Thioredoxin catalyzes the S-nitrosation of the 58. A. Rizk, G. Paul, P. Incardona, M. Bugarski, M. Mansouri, A. Niemann, U. Ziegler, P.
caspase-3 active site cysteine. Nat. Chem. Biol. 1, 154–158 (2005). Berger, I. F. Sbalzarini, Segmentation and quantification of subcellular structures
doi:10.1038/nchembio720 Medline in fluorescence microscopy images using Squassh. Nat. Protoc. 9, 586–596
41. T. Uehara, T. Nakamura, D. Yao, Z.-Q. Shi, Z. Gu, Y. Ma, E. Masliah, Y. Nomura, S. (2014). doi:10.1038/nprot.2014.037 Medline
A. Lipton, S-nitrosylated protein-disulphide isomerase links protein misfolding to 59. B. Spencer, L. Rank, J. Metcalf, P. Desplats, Identification of insulin receptor splice
neurodegeneration. Nature 441, 513–517 (2006). doi:10.1038/nature04782 variant B in neurons by in situ detection in human brain samples. Sci. Rep. 8, 4070
Medline (2018). doi:10.1038/s41598-018-22434-2 Medline
42. L. Mucke, E. Masliah, G.-Q. Yu, M. Mallory, E. M. Rockenstein, G. Tatsuno, K. Hu, D.

Downloaded from http://science.sciencemag.org/ on December 4, 2020


Kholodenko, K. Johnson-Wood, L. McConlogue, High-level neuronal expression of ACKNOWLEDGMENTS
Aβ1–42 in wild-type human amyloid protein precursor transgenic mice: We thank Drs. David Bredt and Solomon Snyder (Johns Hopkins) for HEK-nNOS
Synaptotoxicity without plaque formation. J. Neurosci. 20, 4050–4058 (2000). cells, Traci Fang-Newmeyer (Sanford Burnham Prebys Medical Discovery
doi:10.1523/JNEUROSCI.20-11-04050.2000 Medline Institute) and Weiping Tan (The Scripps Research Institute) for preparing
43. H. Liu, N. Povysheva, M. E. Rose, Z. Mi, J. S. Banton, W. Li, F. Chen, D. P. Reay, G. cultures, and Scott R. McKercher (The Scripps Research Institute) for help with
Barrionuevo, F. Zhang, S. H. Graham, Role of UCHL1 in axonal injury and functional manuscript critical review and preparation. Funding: This work was supported in
recovery after cerebral ischemia. Proc. Natl. Acad. Sci. U.S.A. 116, 4643–4650 part by NIH grants RF1 AG057409, R01 AG056259, R01 DA048882, R01
(2019). doi:10.1073/pnas.1821282116 Medline NS086890, P30 NS076411, P01 ES016738 and DP1 DA041722 (to S.A.L.), R01
44. J. R. Lancaster Jr., How are nitrosothiols formed de novo in vivo? Arch. Biochem. AG061845 (to T.N.), P30 AG062429 and R01 AG018440 (to R.A.R), and P41
Biophys. 617, 137–144 (2017). doi:10.1016/j.abb.2016.10.015 Medline GM103533 (to J.R.Y.); by a Distinguished Investigator Award from the Brain &
45. D. Seth, D. T. Hess, A. Hausladen, L. Wang, Y. J. Wang, J. S. Stamler, A multiplex Behavior Research Foundation (to S.A.L.); by a New Investigator Award from the
enzymatic machinery for cellular protein S-nitrosylation. Mol. Cell 69, 451–464.e6 Alzheimer’s Association (to T.N.); by the Michael J. Fox Foundation (to S.A.L. and
(2018). doi:10.1016/j.molcel.2017.12.025 Medline T.N.); and by the Step Family Foundation (to S.A.L.). Author contributions:
46. M. R. Talipov, Q. K. Timerghazin, Protein control of S-nitrosothiol reactivity: S.A.L. conceived, designed, and supervised the execution of the entire project.
Interplay of antagonistic resonance structures. J. Phys. Chem. B 117, 1827–1837 T.N., C.O., and S.A.L. formulated the detailed research plans, interpreted
(2013). doi:10.1021/jp310664z Medline experimental results, and wrote the manuscript. T.N. and C.O. performed a
47. D. Reznick, M. Bryant, D. Holmes, The evolution of senescence and post- majority of molecular and biochemical experiments, X.Z. carried out biochemical
reproductive lifespan in guppies (Poecilia reticulata). PLOS Biol. 4, e7 (2006). experiments with the J20 mouse model of AD, and A.K.D. and H.S. conducted
doi:10.1371/journal.pbio.0040007 Medline some biochemical experiments with cell culture models. L.L. and J.R.Y. were
48. M. W. Akhtar, S. Sanz-Blasco, N. Dolatabadi, J. Parker, K. Chon, M. S. Lee, W. responsible for mass spectrometry analysis, K.M.L. performed lentiviral
Soussou, S. R. McKercher, R. Ambasudhan, T. Nakamura, S. A. Lipton, Elevated injections into mouse brains, and D.G. prepared lentiviral constructs for the
glucose and oligomeric β-amyloid disrupt synapses via a common pathway of study. B.S., E.M., and R.S. were responsible for histological analysis. Competing
aberrant protein S-nitrosylation. Nat. Commun. 7, 10242 (2016). interests: D.G. is currently Director of Vector Development at LocanaBio, Inc.
doi:10.1038/ncomms10242 Medline (San Diego, California). Data and materials availability: All data are available in
49. K. Hsiao, P. Chapman, S. Nilsen, C. Eckman, Y. Harigaya, S. Younkin, F. Yang, G. the main text or the supplementary materials. All plasmids generated in this
Cole, Correlative memory deficits, Abeta elevation, and amyloid plaques in study are available from Stuart A. Lipton under a material transfer agreement
transgenic mice. Science 274, 99–102 (1996). doi:10.1126/science.274.5284.99 with The Scripps Research Institute.
Medline
50. S. Z. Lei, Z.-H. Pan, S. K. Aggarwal, H.-S. V. Chen, J. Hartman, N. J. Sucher, S. A. SUPPLEMENTARY MATERIALS
Lipton, Effect of nitric oxide production on the redox modulatory site of the NMDA science.sciencemag.org/cgi/content/full/science.aaw0843/DC1
receptor-channel complex. Neuron 8, 1087–1099 (1992). doi:10.1016/0896- Figs. S1 to S7
6273(92)90130-6 Medline Tables S1 and S2
51. M. T. Forrester, J. W. Thompson, M. W. Foster, L. Nogueira, M. A. Moseley, J. S. MDAR Reproducibility Checklist
Stamler, Proteomic analysis of S-nitrosylation and denitrosylation by resin-
assisted capture. Nat. Biotechnol. 27, 557–559 (2009). doi:10.1038/nbt.1545 15 November 2018; accepted 18 November 2020
Medline Published online 3 December 2020
52. M. P. Washburn, D. Wolters, J. R. Yates 3rd, Large-scale analysis of the yeast 10.1126/science.aaw0843
proteome by multidimensional protein identification technology. Nat. Biotechnol.
19, 242–247 (2001). doi:10.1038/85686 Medline
53. J. K. Eng, A. L. McCormack, J. R. Yates 3rd, An approach to correlate tandem mass
spectral data of peptides with amino acid sequences in a protein database. J. Am.
Soc. Mass Spectrom. 5, 976–989 (1994). doi:10.1016/1044-0305(94)80016-2
Medline
54. D. L. Tabb, W. H. McDonald, J. R. Yates 3rd, DTASelect and Contrast: Tools for
assembling and comparing protein identifications from shotgun proteomics. J.
Proteome Res. 1, 21–26 (2002). doi:10.1021/pr015504q Medline

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 10
Downloaded from http://science.sciencemag.org/ on December 4, 2020
Fig. 1. S-Nitrosylation of Uch-L1 at Cys152. (A) SNO-Uch-L1 formation by biotin-switch assay in SH-SY5Y cells
exposed to 100 μM SNOC. As a negative control, ascorbate (Asc) was omitted. Values are expressed as mean
+ SEM (n = 3 biological replicates per group). **P < 0.01 by ANOVA. (B) SNO-Uch-L1 formation by biotin-switch
assay in HEK-nNOS cells exposed to calcium ionophore A23187. (C) Presence of SNO-Uch-L1 in the
cerebrocortex of 3-6 mos-old hAPP-J20 AD transgenic mice by biotin-switch assay. Values are mean + SEM (n
= 6 per group). *P < 0.05 by Student’s t test. (D) S-Nitrosylation of Uch-L1 at Cys152. HEK cells expressing myc-
tagged WT Uch-L1 or cysteine mutants of Uch-L1 were exposed to SNOC. Lack of a band by biotin-switch assay
for Uch-L1(C152S) indicates that it is the predominant site of S-nitrosylation. Values are expressed as mean +
SEM (n = 3-4 per group). *P < 0.05 compared to “WT Uch-L1 + SNOC” by ANOVA. (E) S-Nitrosylation of Uch-
L1 at Cys152 by biotin-switch assay in HEK-nNOS cells. HEK-nNOS cells expressing myc-tagged Uch-L1 WT or
C152S mutant were exposed to A23187 to increase intracellular Ca2+ and thus activate nNOS to generate
endogenous NO. The NOS inhibitor NG-nitro-l-arginine (NNA, 1 mM) was used to inhibit NO generation as a
control. Values are expressed as mean + SEM (n = 3 per group). *P < 0.05 by ANOVA. Uncropped immunoblots
are included in fig. S7.

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 11
Downloaded from http://science.sciencemag.org/ on December 4, 2020
Fig. 2. Mass spectrometry (MS) identification of S-nitrosylated Cys
residue in Uch-L1 and atomic resolution model. (A) Table summarizing S-
nitrosylated peptides for Uch-L1 by MS analysis. Four peptides were
identified containing Cys152, and one peptide with Cys220. Spectral counting
clearly demonstrated that Cys152 is the predominant S-nitrosylation site on
Uch-L1 (total spectral counts 33 for Cys152 vs. 2 for Cys220). (B) Tandem
mass spectrum of a Uchl-L1 peptide identifying S-nitrosylation at Cys152. The
measured mass, charge state, and measurement accuracy of the precursor
ion are displayed. The b (blue) and y (red) fragment ions are annotated with
the measured mass. (C) Crystal structure of Uch-L1 (PDB ID: 2ETL) near
Cys152 (yellow). Magenta: Glu7 and Arg153. Molecular visualization and
graphics handling were performed using PyMol.

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 12
Downloaded from http://science.sciencemag.org/ on December 4, 2020
Fig. 3. Aβ-induced S-nitrosylation of Uch-L1 contributes to AD-related synapse loss. (A) SNO-Uch-L1 formation
in primary rat cerebrocortical neurons in culture after 4.5 hours exposure to 500 nM oligomerized Aβ1-42 assayed by
the biotin-switch method (upper panel). Quantification of biotin-switch blots (lower panel). Values are mean + SEM
(n = 3 per group). *P < 0.05 by Student’s t test. (B and C) Exposure of rat cerebrocortical neurons in vitro to Aβ
oligomers (500 nM) for 24 hours resulted in decreased dendritic spines, visualized by GFP transfection (B).
Transfection with non-nitrosylatable mutant Uch-L1(C152S) abrogated Aβ-induced dendritic spine damage in a
statistically significant manner, while WT Uch-L1 manifested a lesser, nonsignificant effect. Values are mean + SEM
(spines scored for n = 25 neurons). *P < 0.05 by ANOVA with Bonferroni correction. (D and E) Lentiviral expression
of Uch-L1(C152S) in vivo prevents loss of presynaptic marker in hAPP-J20 mice. Representative immunostaining of
hippocampal sections with synaptophysin antibody (SY38; pseudo-colored red). Scale bar, 200 μm (D).
Quantification of hippocampal immunohistochemistry from WT and hAPP-J20 mice injected with WT Uch-L1, mutant
Uch-L1(C152S), or empty lentiviral vectors (E). Immunohistochemistry was performed with synaptophysin antibody
(SY38). Values are mean + SEM (n = 18 mice injected). *P < 0.05, ***P < 0.001 by Student’s t test with Bonferroni
correction.
First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 13
Downloaded from http://science.sciencemag.org/ on December 4, 2020
Fig. 4. 4. Transnitrosylation from SNO-Uch-L1 to Cdk5 on biotin-
switch assays. (A) SNO-Uch-L1 formed 10-min after exposure of HEK-
293 cells to 100 μM SNOC, but not after exposure to control
represented by “old” SNOC from which the NO group had been
dissipated, although this solution might still contain nitrite and
disulfides. Formation of SNO-Cdk5 was also observed, and this was
partially abrogated by siRNA knockdown of Uch-L1 (siUch-L1)
compared to control siRNA (siCTL) cells. (B) Knockdown of Uch-L1
largely prevented formation of SNO-Drp1, which first appeared 25 min
after exposure to SNOC. (C) Quantification of SNO-Cdk5 on biotin-
switch blots. Relative SNO-Cdk5 levels (SNO-protein/Input protein) in
siCTL cells were increased 10 min after exposure to SNOC vs. 25 min
when compared to siUch-L1 cells exposed to SNOC. Values are mean +
SEM (n = 3 per group). ***P < 0.001 by two-tailed Student’s t test. (D)
Similar type of quantification of SNO-Drp1 on biotin-switch blots. In this
case, relative SNO-Drp1 levels were increased 25 min after exposure to
SNOC vs. 10 min. Values are mean + SEM (n = 3 per group). ***P <
0.001 by two-tailed Student’s t test.

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 14
Fig. 5. Triple transnitrosylation from
SNO-Uch-L1 to SNO-Cdk5 to SNO-
Drp1. (A) Schematic diagram of in
vitro transnitrosylation assay. (B)
Transnitrosylation occurs from WT
SNO-Uch-L1 to Cdk5 to Drp1 but
significantly less with mutant Uch-
L1(C152S). WT Uch-L1-V5 or mutant
Uch-L1(C152S)-V5 from SH-SY5Y
cell lysates was immunoprecipitated
with anti-V5 antibody and exposed to
100 μM SNOC. These
immunoprecipitates were then added
to whole cell lysates expressing HA-
Cdk5, which were subjected to the
biotin-switch assay to assess

Downloaded from http://science.sciencemag.org/ on December 4, 2020


transnitrosylation. Compilation of
representative blots shown. (C and
D) Quantification of Uch-L1 and Cdk5
biotin-switch blots. Values are mean
+ SEM (n = 4 per group). ***P < 0.001
by two-tailed Student’s t test, **P <
0.01, *P < 0.05 by ANOVA. (E)
Quantification of Drp1 biotin-switch
blots. Values are mean + SEM (n = 3
per group). *P < 0.05, **P < 0.01 by
ANOVA. (F) Schematic diagram of in
vitro transnitrosylation assay after
immunoprecipitation to decrease the
amount of endogenous Cdk5. (G)
SNO-Uch-L1 nitrosylation of Drp1 is
affected by the level of endogenous
Cdk5. WT Uch-L1-V5 or Uch-
L1(C152S)-V5 from SH-SY5Y cell
lysates was immunoprecipitated with
anti-V5 antibody, and the
immunoprecipitates exposed to 100
μM SNOC or old SNOC. The
immunoprecipitates containing
SNOC-exposed Uch-L1 proteins were
then incubated with SH-SY5Y cell
lysates expressing Drp1-HA after
substantial immunodepletion of
endogenous Cdk5 with anti-Cdk5
antibody (or IgG control antibody).
Samples were subjected to biotin-
switch assay to assess
transnitrosylation. (H and I)
Quantification of Uch-L1and Drp1
biotin-switch blots. Values are mean
+ SEM (n = 3 per group). **P < 0.01,
*P < 0.05 by ANOVA.

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 15
Downloaded from http://science.sciencemag.org/ on December 4, 2020
Fig. 6. Redox blot quantifying transnitrosylation from Uch-L1 to Cdk5. (A)
Transnitrosylation occurs predominantly from SNO-Uch-L1 to Cdk5 to Drp1
rather than in the opposite direction from SNO-Cdk5 to Uch-L1. Expression of
Uch-L1-V5 and Cdk5-HA increased the formation of SNO-Drp1, as evidenced
on biotin-switch assay. Lysates of SH-SY5Y cells were prepared by V5- or HA-
immunoprecipitation and exposed to 100 μM SNOC. These
immunoprecipitates were then added to whole-cell lysates expressing either
HA-Cdk5 or Uch-L1-V5, which were subjected to the biotin-switch assay to
assess transnitrosylation. (B) SH-SY5Y cells were transfected with Uchl-L1-V5
and Cdk5-HA tagged constructs, and exposed to 50 μM SNOC at room
temperature. After 30 min, cell lysates were prepared, incubated to achieve
steady state (~1 hour, by which time SNOC, a short-lived NO donor, had
completely dissipated), and then subjected to the biotin-switch assay. Methyl-
methanethiosulfonate (MMTS, 25 mM) was used to block free thiols during
the assay for S-nitrosylated protein. Chemically-reduced thiol protein
represents the relative amount of total protein obtained by the biotin-switch
assay performed in the absence of MMTS (w/o MMTS). SNO-protein
represents the relative amount of S-nitrosylated protein obtained by the
biotin-switch assay. The relative concentration of protein in a redox pair in the
S-nitrosylated (oxidized) form and the reduced form can be measured by
quantitative densitometry of their respective bands on the gels (n = 4).

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 16
Downloaded from http://science.sciencemag.org/ on December 4, 2020
Fig. 7. S-Nitrosylation of Uch-L1 in human AD brains and schema of transnitrosylation in the
pathophysiology of synapse loss in AD. (A) Human brain tissues from control or AD patients were
subjected to the biotin-switch assay to detect SNO-Uch-L1. (B) Quantification of the ratio of SNO-
Uch-L1 to total Uch-L1 in human brains and in cell-based assays. Biotin-switch assays and
immunoblot analyses were quantified by densitometry, and the relative ratio of SNO-Uch-L1 to total
Uch-L1 was calculated for the following conditions: In vitro in cerebrocortical neurons after Aβ
exposure (as shown in Fig. 3A), in vivo in the hAPP-J20 mouse model of AD (as shown in Fig. 1C),
and in human AD brains vs. control human brains. Values are mean + SEM (n = 6 for control human
brains, n = 7 samples from 5 AD human brains, n = 3 for each group in primary neuron experiments,
n = 6 for each group in mouse brain experiments). ***P < 0.001, *P < 0.05 by Student’s t test. (C)
Biochemical schema of transnitrosylation pathway leading to synaptic damage and consequent
memory loss in AD. Note that these transnitrosylation reactions may be direct or indirect, with
additional, as yet unknown, members of the transnitrosylation network still to be discovered.

First release: 3 December 2020 www.sciencemag.org (Page numbers not final at time of first release) 17
Noncanonical transnitrosylation network contributes to synapse loss in Alzheimer's disease
Tomohiro Nakamura, Chang-ki Oh, Lujian Liao, Xu Zhang, Kevin M. Lopez, Daniel Gibbs, Amanda K. Deal, Henry R. Scott, Brian
Spencer, Eliezer Masliah, Robert A. Rissman, John R. Yates III and Stuart A. Lipton

published online December 3, 2020

Downloaded from http://science.sciencemag.org/ on December 4, 2020


ARTICLE TOOLS http://science.sciencemag.org/content/early/2020/12/02/science.aaw0843

SUPPLEMENTARY http://science.sciencemag.org/content/suppl/2020/12/02/science.aaw0843.DC1
MATERIALS

REFERENCES This article cites 59 articles, 20 of which you can access for free
http://science.sciencemag.org/content/early/2020/12/02/science.aaw0843#BIBL

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Use of this article is subject to the Terms of Service

Science (print ISSN 0036-8075; online ISSN 1095-9203) is published by the American Association for the Advancement of
Science, 1200 New York Avenue NW, Washington, DC 20005. The title Science is a registered trademark of AAAS.
Copyright © 2020, American Association for the Advancement of Science

You might also like